Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Microbiol Biotechnol ; 31(5): 667-675, 2021 May 28.
Article in English | MEDLINE | ID: mdl-33879639

ABSTRACT

Streptococcus agalactiae is an important bacterial pathogen and causative agent of diseases including neonatal sepsis and meningitis, as well as infections in healthy adults and pregnant women. Although antibiotic treatments effectively relieve symptoms, the emergence and transmission of multidrug-resistant strains indicate the need for an effective immunotherapy. Effector T helper (Th) 17 cells are a relatively newly discovered subpopulation of helper CD4+ T lymphocytes, and which, by expressing interleukin (IL)-17A, play crucial roles in host defenses against a variety of pathogens, including bacteria and viruses. However, whether S. agalactiae infection can induce the differentiation of CD4+ T cells into Th17 cells, and whether IL-17A can play an effective role against S. agalactiae infections, are still unclear. In this study, we analyzed the responses of CD4+ T cells and their defensive effects after S. agalactiae infection. The results showed that S. agalactiae infection induces not only the formation of Th1 cells expressing interferon (IFN)-γ, but also the differentiation of mouse splenic CD4+ T cells into Th17 cells, which highly express IL-17A. In addition, the bacterial load of S. agalactiae was significantly increased and decreased in organs as determined by antibody neutralization and IL-17A addition experiments, respectively. The results confirmed that IL-17A is required by the host to defend against S. agalactiae and that it plays an important role in effectively eliminating S. agalactiae. Our findings therefore prompt us to adopt effective methods to regulate the expression of IL-17A as a potent strategy for the prevention and treatment of S. agalactiae infection.


Subject(s)
Interleukin-17/immunology , Streptococcal Infections/immunology , Streptococcus agalactiae/physiology , Th17 Cells/immunology , Animals , Bacterial Load/drug effects , Bacterial Load/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Cytokines/immunology , Female , Interleukin-17/administration & dosage , Interleukin-17/antagonists & inhibitors , Mice , Spleen/immunology , Spleen/microbiology , Streptococcal Infections/microbiology , Th1 Cells/immunology
2.
Front Microbiol ; 11: 556, 2020.
Article in English | MEDLINE | ID: mdl-32390957

ABSTRACT

ATP-binding cassette transporters are ubiquitous in almost all organisms. The Escherichia coli genome is predicted to encode 69 ABC transporters. Eleven of the ABC transporters are presumed to be exporters, of which seven are possible drug export transporters. There has been minimal research on the function of YbhFSR, which is one of the putative drug resistance exporters. In this study, the ybhF gene of this transporter was characterized. Overexpression and knockout strains of ybhF were constructed. The ATPase activity of YbhF was studied using the malachite green assay, and the efflux abilities of knockout strains were demonstrated by using ethidium bromide (EB) as a substrate. The substrates of YbhFSR efflux, examined with the minimum inhibitory concentration (MIC), were determined to be tetracycline, oxytetracycline, chlortetracycline, doxycycline, EB, and Hoechst33342. Furthermore, tetracycline and EB efflux and accumulation experiments confirmed that the substrates of YbhFSR were tetracyclines and EB. The MIC assay and the fluorescence test results showed that tetracyclines are likely to be the major antibiotic substrate of YbhFSR. The existence of the signature NatA motif suggested that YbhFSR may also function as a Na+/H+ transporter. Overexpression of YbhF in E. coli KNabc lacking crucial Na+/H+ transporters conferred tolerance to NaCl, LiCl, and an alkaline pH. Together, the results showed that YbhFSR exhibited dual functions as a drug efflux pump and a Na+ (Li+)/H+ antiporter.

3.
J Microbiol Biotechnol ; 30(7): 982-995, 2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32347079

ABSTRACT

A putative multidrug efflux gene, yddA, was cloned from the Escherichia coli K-12 strain. A drugsensitive strain of E. coli missing the main multidrug efflux pump AcrB was constructed as a host and the yddA gene was knocked out in wild-type (WT) and drug-sensitive E. coliΔacrB to study the yddA function. Sensitivity to different substrates of WT E.coli, E. coliΔyddA, E. coliΔacrB and E. coliΔacrBΔyddA strains was compared with minimal inhibitory concentration (MIC) assays and fluorescence tests. MIC assay and fluorescence test results showed that YddA protein was a multidrug efflux pump that exported multiple substrates. Three inhibitors, ortho-vanadate, carbonyl cyanide m-chlorophenylhydrazone (CCCP), and reserpine, were used in fluorescence tests. Ortho-vanadate and reserpine significantly inhibited the efflux and increased accumulation of ethidium bromide and norfloxacin, while CCCP had no significant effect on YddA-regulated efflux. The results indicated that YddA relies on energy released from ATP hydrolysis to transfer the substrates and YddA is an ABC-type multidrug exporter. Functional study of unknown ATP-binding cassette (ABC) superfamily transporters in the model organism E. coli is conducive to discovering new multidrug resistance-reversal targets and providing references for studying other ABC proteins of unknown function.


Subject(s)
Cloning, Molecular/methods , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/genetics , Anti-Bacterial Agents/pharmacology , Carbonyl Cyanide m-Chlorophenyl Hydrazone , Escherichia coli K12/genetics , Genes, MDR , Microbial Sensitivity Tests , Multidrug Resistance-Associated Proteins , Norfloxacin/pharmacology , Reserpine/pharmacology
4.
Microb Pathog ; 136: 103676, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31437577

ABSTRACT

The purpose of this investigation was to construct a recombinant Escherichia coli strain displaying the Staphylococcus aureus target of RNAIII activating protein (TRAP) on its surface, and to investigate the strain for its immunogenicity. The lpp'ompA and lpp'ompA-TRAP genes were fused by the overlap polymerase chain reaction and then ligated into expression plasmid pQE30 producing pLO and pLO-TRAP. These two recombinant plasmids were transformed into E. coli XL1-Blue, resulting in XL1-Blue/pLO and XL1-Blue/pLO-TRAP, which were induced to express protein. The expressed TRAP protein was displayed on the surface of XL1-Blue as judged by whole cell ELISA, flow cytometric analysis, and laser scanning confocal microscopy using the lpp'ompA surface display system. ICR mice were intramuscularly immunized with recombinant strains XL1-Blue/pLO and XL1-Blue/pLO-TRAP as well as recombinant protein TRAP. Immunized mice were assessed for anti-TRAP antibody and lymphocytes for secreted IL-4 and IFN-γ by ELISPOT and secreted IL-17A by indirect ELISA. Immunized mice were challenged with S. aureus Newman and HLJ23-1 strains. The results showed both XL1-Blue/pLO-TRAP and TRAP protein immunized mice to produce better cellular and humoral immunity than XL1-Blue/pLO and PBS injected mice.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Cell Surface Display Techniques , Membrane Proteins/immunology , Recombinant Fusion Proteins/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Cytokines/metabolism , Disease Models, Animal , Drug Carriers , Enzyme-Linked Immunospot Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Injections, Intramuscular , Lymphocytes/immunology , Membrane Proteins/genetics , Mice, Inbred ICR , Recombinant Fusion Proteins/genetics , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics
5.
Microb Pathog ; 118: 39-47, 2018 May.
Article in English | MEDLINE | ID: mdl-29522802

ABSTRACT

The GapC protein of Staphylococcus aureus (S. aureus) is a surface protein that is highly conserved among Staphylococcus strains, and it can induce protective humoral immune responses. However, B-cell epitopes in S. aureus GapC have not been reported. In this study, we generated a monoclonal antibody (mAb2A9) targeting S. aureus GapC. Through a passive immunity test, mAb2A9 was shown to partially protect mice against S. aureus infection. We screened the motif 236PVATGSLTE243 that is recognized by mAb2A9 using a phage-display system. The motif sequence exactly matched amino acids 236-243 of the S. aureus GapC protein. Then, we identified the key amino acids in the motif using site-directed mutagenesis. Site-directed mutagenesis revealed that residues P236, G240, L242, and T243 formed the core of the 236PVATGSLT243 motif. In addition, this epitope was proven to be located on the surface of S. aureus, and it induced a protective humoral immune response against S. aureus infection in immunized mice. Overall, our results characterized a conserved B-cell epitope, which will be an attractive target for designing effective epitope-based vaccines against S. aureus infection.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/metabolism , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Vaccines , Bacteriophages , Cell Surface Display Techniques , Disease Models, Animal , Epitopes/chemistry , Epitopes/immunology , Female , Immunity , Immunization, Passive , Macrophages/immunology , Mice , Mice, Inbred BALB C , Models, Molecular , Mutagenesis, Site-Directed , Phagocytosis , Protein Conformation , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Sequence Alignment , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/genetics
6.
Sci Rep ; 8(1): 3580, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483570

ABSTRACT

Staphylococcus aureus can cause different types of diseases from mild skin infections to life-threatening sepsis worldwide. Owing to the emergence and transmission of multidrug-resistant strains, developing an impactful immunotherapy especially vaccine control approach against S. aureus infections is increasingly encouraged and supported. S. aureus manganese transport protein C (MntC), which is a highly-conserved cell surface protein, can elicit protective immunity against S. aureus and Staphylococcus epidermidis. In this study, we evaluated the humoral immune response and CD4+ T cell-mediated immune responses in a mouse peritonitis model. The results showed that MntC-specific antibodies conferred an essential protection for mice to reduce invasion of S. aureus, which was corroborated via the opsonophagocytic killing assay and passive immunization experiment in mice, and moreover MntC-induced Th17 played a remarkable part in preventing S. aureus infection since the MntC-induced protective immunity decreased after neutralization of IL-17 by antibody in vivo and the Th17 adoptive transferred-mice could partly resist S. aureus challenge. In conclusion, we considered that the MntC-specific antibodies and MntC-specific Th17 cells play cooperative roles in the prevention of S. aureus infection.


Subject(s)
Bacterial Proteins/immunology , Peritonitis/immunology , Peritonitis/microbiology , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Th17 Cells/immunology , Animals , Antibodies, Bacterial , Cell Proliferation/drug effects , Disease Models, Animal , Female , Immunity, Cellular , Immunity, Humoral , Immunization, Passive/psychology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Manganese/metabolism , Mice , Mice, Inbred BALB C
7.
PLoS One ; 13(1): e0190452, 2018.
Article in English | MEDLINE | ID: mdl-29304128

ABSTRACT

The impact of epidemic Staphylococcus aureus (S. aureus) on public health is increasing. Because of the abuse of antibiotics, the antibiotic resistance of S. aureus is increasing. Thus, there is an urgent need to develop new immunotherapies and immunoprophylaxes. Previous studies showed that the GapC protein of S. aureus, which is a surface protein with high glyceraldehyde 3-phosphate dehydrogenase activity, transferrin binding activity, and other biological activities, is highly conserved. GapC induces an effective humoral immune response in vivo. However, the B-cell epitopes of S. aureus GapC have not been well identified. Here we used the bioinformatics tools to analyze the sequence of GapC, and we generated protective anti-GapC monoclonal antibodies (mAbs). A protective mAb (1F4) showed strong specificity to GapC and the ability to induce macrophages to phagocytose S. aureus. We screened the motif 272GYTEDEIVSSD282, which was recognized by mAb 1F4, using a phage display system. Then, we used site-directed mutagenesis to identify key amino acids in the motif. Residues G272 D276 E277 I278 and V279 formed the core of the 272GYTEDEIVSSD282 motif. In addition, we showed that this epitope peptide induced a protective humoral immune response against S. aureus infection in immunized mice. Our results will be useful for the further study of epitope-based vaccines against S. aureus infection.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , B-Lymphocytes/immunology , Bacterial Proteins/immunology , Bacteriophages/genetics , Epitopes/immunology , Peptide Library , Amino Acid Sequence , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Monoclonal/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Phagocytosis , Protein Structure, Tertiary
8.
Cytogenet Genome Res ; 153(1): 36-45, 2017.
Article in English | MEDLINE | ID: mdl-29169149

ABSTRACT

Interferon-γ (IFN-γ), a cytokine produced by activated natural killer cells and T lymphocytes, is an important regulator of innate and adaptive immunity. Interleukin (IL)-18, also known as IFN-γ-inducing factor, is a cytokine that induces T and natural killer cells to produce IFN-γ. In this study, the chicken IL-18 (ChIL-18) and chicken IFN-γ (ChIFN-γ) genes were inserted into the pET28a prokaryotic expression vector, resulting in pET28a-IL-18 and pET28a-IFN-γ, respectively. These plasmids were transformed into Escherichia coli strain BL21, and the ChIL-18 and ChIFN-γ proteins were expressed and purified. To determine their antiviral activities, 200 ng/mL of ChIL-18 and/or ChIFN-γ were inoculated into chicken embryonic fibroblast cells. After 24 h, one 50% tissue culture infective dose (TCID50) of infectious bursal disease virus (IBDV) was inoculated into the chicken embryonic fibroblast cells. The results showed that the antiviral effect of ChIL-18 and ChIFN-γ in combination was better than that of ChIL-18 or ChIFN-γ alone. Next, 14-day-old chicken were injected with 200 µg of ChIL-18 and/or ChIFN-γ and then were challenged with 103 TCID50 of IBDV via intraperitoneal injection. The results showed that the proliferation of IBDV was inhibited by the injection of the recombinant proteins, especially the combination of ChIL-18 and ChIFN-γ, as evidenced by cytokine detection, quantitative PCR, and pathology analyses. These results indicate that ChIL-18 and ChIFN-γ could inhibit IBDV infection and the combination of ChIL-18 and ChIFN-γ has a better inhibitory effect than either cytokine alone.


Subject(s)
Birnaviridae Infections/prevention & control , Infectious bursal disease virus/immunology , Interferon-gamma/genetics , Interleukin-18/genetics , Virus Replication/immunology , Animals , Antiviral Agents/metabolism , Birnaviridae Infections/immunology , Birnaviridae Infections/virology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Chick Embryo , Chickens , Escherichia coli/genetics , Escherichia coli/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-18/biosynthesis , Interleukin-18/immunology , Killer Cells, Natural/immunology , Macrophage Activation/immunology , Macrophages/immunology , Plasmids/genetics , Virus Replication/genetics
9.
Microb Pathog ; 112: 30-37, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28942173

ABSTRACT

Manganese transport protein C (MntC) of Staphylococcus aureus represents an excellent vaccine-candidate antigen. The important role of CD4+ T cells in effective immunity against S. aureus infection was shown; however, CD4+ T cell-specific epitopes on S. aureus MntC have not been well identified. Here, we used bioinformatics prediction algorithms to evaluate and identify nine candidate epitopes within MntC. Our results showed that peptide M8 emulsified in Freund's adjuvant induced a much higher cell-proliferation rate as compared with controls. Additionally, CD4+ T cells stimulated with peptide M8 secreted significantly higher levels of interferon-γ and interleukin-17A. These results suggested that peptide M8 represented an H-2d (I-E)-restricted Th17-specific epitope.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/isolation & purification , Manganese/metabolism , Protein C/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Algorithms , Amino Acid Sequence , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Cytokines/metabolism , Epitope Mapping , Escherichia coli/genetics , Female , Interferon-gamma/metabolism , Interleukin-17/metabolism , Major Histocompatibility Complex , Mice , Mice, Inbred BALB C , Protein C/genetics , Protein C/immunology , Protein Structure, Secondary , Recombinant Proteins/immunology , Staphylococcal Infections/immunology , Th1 Cells/immunology , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...